The isolation of CHO cells with a site conferring a high and reproducible transgene amplification rate

https://doi.org/10.1016/j.jbiotec.2013.01.016Get rights and content

Abstract

Co-amplification of transgenes using the dihydrofolate reductase/methotrexate (DHFR/MTX) system is a widely used method for the isolation of Chinese hamster ovary (CHO) cell lines that secrete high levels of recombinant proteins. A bottleneck in this process is the stepwise selection for MTX resistant populations; which can be slow, tedious and erratic. We sought to speed up and regularize this process by isolating dhfr CHO cell lines capable of integrating a transgene of interest into a defined chromosomal location that supports a high rate of gene amplification. We isolated 100 independent transfectants carrying a gene for human adenosine deaminase (ada) linked to a φC31 attP site and a portion of the dihydrofolate reductase (dhfr) gene. Measurement of the ada amplification rate in each transfectant using Luria–Delbruck fluctuation analysis revealed a wide clonal variation; sub-cloning showed these rates to be heritable. Site directed recombination was used to insert a transgene carrying a reporter gene for secreted embryonic alkaline phosphatase (SEAP) as well as the remainder of the dhfr gene into the attP site at this location in several of these clones. Subsequent selection for gene amplification of the reconstructed dhfr gene in a high ada amplification candidate clone (DG44-HA-4) yielded reproducible rates of seap gene amplification and concomitant increased levels of SEAP secretion. In contrast, random integrations of the dhfr gene into clone HA-4 did not yield these high levels of amplification. This cell line as well as this method of screening for high amplification rates may prove helpful for the reliable amplification of recombinant genes for therapeutically or diagnostically useful proteins.

Highlights

► We measured the rate of gene amplification in 100 CHO cell transfectant clones. ► Amplification rates were heritable in subclones. ► Clones with a high rate were targeted for insertion of a reporter gene. ► The reporter gene could be amplified at a high rate in several clones. ► This method could be used for more rapid and reliable gene amplification.

Introduction

The phenomenon of gene amplification is often exploited to produce therapeutic recombinant proteins, monoclonal antibodies being the prime example. A gene or genes coding for the therapeutic protein(s) of choice (e.g., monoclonal antibody heavy and light chain) are transfected into a host cell along with a selectable drug-resistance marker gene. After an initial isolation of transfectants exhibiting a minimal level of drug resistance, selection for transfectants that have acquired a modestly higher level of resistance by virtue of amplification of the marker gene is carried out. Many iterations of this selection process can result in cell clones with a high level of resistance, a high level of marker gene expression, and a high number of copies of the marker gene. Since the size of the amplified region is much larger than a single gene (Hamlin, 1992) and since transfected genes usually co-integrate in the host genome, even when co-transfected on separate plasmids (Chen and Chasin, 1998), the gene of interest is also co-amplified and its protein produced at a high level (Wigler et al., 1980).

A popular system for this approach uses a gene specifying the enzyme dihydrofolate reductase (DHFR) as the selective marker (Ringold et al., 1981), methotrexate (MTX) as the drug and a Chinese hamster ovary (CHO) cell line deficient in this ubiquitous enzyme (Urlaub and Chasin, 1980, Urlaub et al., 1983, Jayapal et al., 2007) as a host. CHO cells are capable of gene amplification, a trait not shared by normal cells (Livingstone et al., 1992, Wahl et al., 1984a, Yin et al., 1992). In this case the target of the drug is DHFR, and resistance is often gained by overproducing it. Transfected cells are cultured in a medium lacking a source of purine and thymidine nucleotides. Since DHFR-deficient host cells are unable to synthesize these metabolites only the transfectants can grow. MTX is a specific and tight-binding inhibitor of DHFR, but amplified cells that have undergone dhfr amplification (“amplificants”) can overcome a judiciously chosen concentration of the drug.

Although the dhfr/MTX gene amplification method can result in as much as a 1000-fold increase in gene copy number (Kaufman and Sharp, 1982), it suffers from long and variable development times. Each amplification step brings about only a modest increase in gene copy number, thus severely limiting the concentration of MTX that can be applied at each step. It is common to take six months or longer to isolate a cell line with the desired recombinant protein production level. This time bottleneck inhibits the rapid testing of multiple new candidates for pre-clinical evaluation and ultimately limits how quickly a new drug candidate gets to market (Trill et al., 1995). Use of MTX amplification may also have unforeseen effects on CHO cells since chromosomal translocations often accompany gene amplification. Additionally, amplified genes are not always stable, resulting in a decrease in gene copy number unless selective pressure is maintained. Due to these drawbacks, this approach is often not used despite its potential to yield high gene copy number clones. Industry has reacted by using novel promoter-enhancer and chromatin opening elements and developing high-throughput techniques to isolate rare high producing clones. Despite these drawbacks, regularizing this process and decreasing the time to implement it could lead to its more widespread use in the development of useful biologics.

Although the factors influencing gene amplification are not well understood, it has been shown that clones within a CHO transfectant population exhibit a wide distribution in the yield of amplificants (Kito et al., 2002, Wahl et al., 1984b). The simplest explanation for this variability is a position effect within the CHO genome. We reasoned that the gene amplification regimen could be both shortened and made more reliable by inserting the gene of interest along with the dhfr gene at a defined locus in the CHO genome chosen for supporting a high amplification rate. We identified such sites by screening 100 transfectants for their rate of amplification (as opposed to the frequency of amplified cells) using Luria–Delbruck fluctuation analysis. A chosen site could subsequently be used to insert genes of interest via an included site specific recombination sequence. A winning clone was shown to amplify a dhfr gene along with a gene of interest at a rate higher than randomly integrated transfectants and to secrete a protein of interest at increased levels. This clone or similarly isolated clones may prove advantageous for those using gene amplification in CHO cells to drive high level production of therapeutically or diagnostically useful proteins.

Section snippets

Plasmid construction

We constructed a vector (pKAPD1) to be transfected into host DHFR-deficient DG44 cells (Urlaub et al., 1983, Urlaub et al., 1986) carrying a neomycin resistance gene and an amplifiable human adenosine deaminase (ada) gene. A portion of the dhfr gene and a φC31 attP site were included for subsequent recombination and amplification steps. A fragment containing the dhfr promoter, exon 1 and part of intron 1 was isolated by PCR using primers appended with AscI restriction site ends (forward:

Strategy

Our goal was to isolate a CHO DG44 cell with a site specific recombination site at a location where gene amplification occurs at a relatively high and reproducible rate. Our plan was to transfect DG44 cells with a plasmid carrying a site specific recombination site linked to a gene that can act as a selective marker for gene amplification. One hundred independent transfectant clones, each presumed to carry this construct at a different location, would be isolated. The gene amplification rate of

Discussion

Our intention was to isolate a clone with a site-specific recombination site located at a chromosomal position that yields a high rate of gene amplification. Such a clone could prove advantageous for the production of a recombinant protein by inserting the corresponding gene into that location. Previous studies (Gajduskova et al., 2007, Kito et al., 2002, Wahl et al., 1984b) showed that only a minority of cell transfectants (27 of 82, 2 of 16, and 1 of 4 respectively) are capable of efficient

Acknowledgments

We would like to thank Mauricio Arias, Shengdong Ke, Vincent Anquetil, Laurens Moore van Tienen, Mrinalini Gururaj, Dennis Weiss, Ron Gejman, Ashira Lubkin and Ye Jung Ferrabolli for insightful advice pertaining to experiments performed throughout this work. This work was funded by ImClone Systems, a wholly owned subsidiary of Eli Lilly and Company.

References (34)

  • H. Zhou et al.

    Generation of stable cell lines by site specific integration of transgenes into engineered Chinese hamster ovary strains using an FLP-FRT system

    Journal of Biotechnology

    (2010)
  • D.S. Allison et al.

    Rapid development of CHO cell lines for high-level production of recombinant antibodies

    BioProcessing Journal

    (2003)
  • L.A. Chasin

    The effect of ploidy on chemical mutagenesis in cultured Chinese hamster cells

    Journal of Cell Physiology

    (1973)
  • C. Chen et al.

    Cointegration of DNA molecules introduced into mammalian cells by electroporation

    Somatic Cell and Molecular Genetics

    (1998)
  • I.T. Chen et al.

    Direct selection for mutations affecting specific splice sites in a hamster dihydrofolate reductase minigene

    Molecular Cell Biology

    (1993)
  • J.J. de Poorter et al.

    Optimization of short-term transgene expression by sodium butyrate and ubiquitous chromatin opening elements (UCOEs)

    Journal of Gene Medicine

    (2007)
  • W.F. Flintoff et al.

    Isolation and partial characterization of three methotrexate-resistant phenotypes from Chinese hamster ovary cells

    Somatic Cell Genetics

    (1976)
  • Cited by (9)

    • How to train your cell - Towards controlling phenotypes by harnessing the epigenome of Chinese hamster ovary production cell lines

      2022, Biotechnology Advances
      Citation Excerpt :

      Thus, identifying regions with high transcriptional activity, accessible chromatin and low nucleosome occupancy should allow a strategy for systematic engineering of stable and high-producing CHO cell lines by integrating the GOI in the desired genomic location resulting in an improved, more stable production phenotype (Dhiman et al., 2020; Balasubramanian et al., 2016; Matasci et al., 2011; O'Brien et al., 2018; Veith et al., 2016; Koduri et al., 2001). By using site directed integration, various producing CHO cell lines were generated, many of which were reported to be stable (Qiao et al., 2009; Baumann et al., 2017; Kawabe et al., 2017; O'Brien et al., 2018; Wang et al., 2017; Grav et al., 2018; Soo and Lee, 2008; Cacciatore et al., 2013; Zhao et al., 2018; Sergeeva et al., 2020). However, GOI expression after targeted integration into loci with high transcriptional activity is still widely varying (Zhao et al., 2018) and although reported results appear promising, further investigation and methodical studies will be necessary to identify the most effective integration sites.

    • Better and faster: Improvements and optimization for mammalian recombinant protein production

      2014, Current Opinion in Structural Biology
      Citation Excerpt :

      Selection and amplification via this system has enabled the production of gram levels of protein (often antibodies) per liter in CHO cells, and improvements are being developed to increase the rapidity of amplification. Strategies to increase the speed of gene amplification include weakening the selection marker (such as via fusion of the murine ornithine decarboxylase PEST protease targeting sequence) and the use of DNA sequences that lead to spontaneous gene amplification in transformed cells (such as the cassette of the mammalian replication initiation region (IR) matrix attachment region (MAR)) [42•,43–45]. In highly analogous systems, selection by the introduction of the glutamate synthase gene into cells that are deficient in this vital enzyme, or rendered so by the inhibition using methionine sulfoximine, is frequently used for protein production (maximal antibody yields of 5 g/l, with a 20–25 weeks timeline from cloning to large-scale fermentation) and is offered commercially by Lonza, Inc. [46].

    • Optimization of Protein Expression in Mammalian Cells

      2019, Current Protocols in Protein Science
    • CCR8<sup>+</sup>FOXp3<sup>+</sup> T<inf>reg</inf> cells as master drivers of immune regulation

      2017, Proceedings of the National Academy of Sciences of the United States of America
    • Cell line development for biomanufacturing processes

      2017, Applied Bioengineering: Innovations and Future Directions
    View all citing articles on Scopus
    View full text